Language selection

Search

Patent 3051314 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3051314
(54) English Title: IMPROVED METHOD FOR EPIGENETIC IMMUNE CELL COUNTING
(54) French Title: PROCEDE AMELIORE DE COMPTAGE EPIGENETIQUE DE CELLULES IMMUNITAIRES
Status: Examination
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12Q 01/6851 (2018.01)
  • C12Q 01/6881 (2018.01)
(72) Inventors :
  • OLEK, SVEN (Germany)
(73) Owners :
  • PRECISION FOR MEDICINE GMBH
(71) Applicants :
  • PRECISION FOR MEDICINE GMBH (Germany)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2018-02-08
(87) Open to Public Inspection: 2018-08-16
Examination requested: 2022-03-01
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/EP2018/053206
(87) International Publication Number: EP2018053206
(85) National Entry: 2019-07-23

(30) Application Priority Data:
Application No. Country/Territory Date
17155496.7 (European Patent Office (EPO)) 2017-02-09

Abstracts

English Abstract

The present invention relates to improved methods for epigenetic blood and immune cell counting, and respective uses and kits.


French Abstract

La présente invention concerne des procédés améliorés de comptage épigénétique de cellules sanguines et immunitaires et des utilisations et des nécessaires respectifs.

Claims

Note: Claims are shown in the official language in which they were submitted.


27
Claims
1 . A method for a quantitative methylation assay for blood immune cells,
comprising the steps
o f
a) providing a defined volume of a sample of human blood comprising diploid
genomic DNA
of blood immune cells to be quantitated;
b) providing an in silico bisulfite-converted recombinant nucleic acid
comprising at least one
demethylation standard gene, and a sequence inversing all CpG dinucleotides to
GpC of said
at least one demethylation standard gene ("standard I");
c) providing a recombinant nucleic acid comprising the demethylated genomic
sequence of
said at least one demethylation standard gene of b), and of said a sequence
inversing all CpG
dinucleotides to GpC of said at least one demethylation standard gene of b)
("calibrator I");
d) providing a recombinant nucleic acid comprising the sequence inversing all
CpG
dinucleotides to GpC of said at least one demethylation standard gene of b)
("spiker I");
e) adding a defined amount of said recombinant nucleic acid of d) to said
sample of a)
("spiking");
f) treating said diploid genomic DNA of the cells to be quantitated of a) and
said recombinant
nucleic acids of c) and d) with bisulfite to convert unmethylated cytosines
into uracil;
g) amplifying of said nucleic acid molecules of a), b), c), and f) using
suitable primer pairs to
produce amplicons; and
h) identifying the blood immune cells (BIC) per volume of sample based on
analyzing said
amplicons.
2. A method for a quantitative methylation assay for blood cells, comprising
the steps of
a) providing a sample of human blood comprising diploid genomic DNA of blood
cells to be
quantitated;
b) providing an in silico bisulfite-converted recombinant nucleic acid
comprising at least one
demethylation standard gene, and at least one blood cell specific gene
("standard II");
c) providing a recombinant nucleic acid comprising the demethylated genomic
sequence of
said at least one demethylation standard gene of b), and of said at least one
blood cell specific
gene of b) ("calibrator II");
d) treating said diploid genomic DNA of the cells to be quantitated of a) and
said recombinant
nucleic acid of c) with bisulfite to convert unmethylated cytosines into
uracil;

28
e) amplifying of said nucleic acid molecules of a), b), c), and d) using
suitable primer pairs to
produce amplicons; and
f) identifying the percent of demethylation per all cells (DDC) based on
analyzing said
amplicons.
3. A method for determining the absolute copy number of methylated genes of an
immune
cell type, comprising
a) Performing the method according to claim 1,
b) Performing the method according to claim 2, and
c) Multiplying the BIC as identified with the DDC as identified.
4. The method according to any one of claims 1 to 3, wherein said blood immune
cell is
selected from a leukocyte, a T-lymphocyte, a granulocyte, a monocyte, a B-
lymphocyte
and/or an NK-cell.
5. The method according to any one of claims 1 to 4, wherein said recombinant
nucleic acid
molecule is selected from a plasmid, a yeast artificial chromosome (YAC),
human artificial
chromosome (HAC), PI-derived artificial chromosome (PAC), a bacterial
artificial
chromosome (BAC), and a PCR-product.
6. The method according to any one of claims 1 to 5, wherein said
demethylation standard
gene is selected from a gene expressed in all cells to be detected, such as,
for example, a
housekeeping gene, such as, for example, GAPDH.
7. The method according to any one of claims 1 to 6, wherein said blood cell
specific gene is
selected from a gene expressed in all blood cells to be detected, such as, for
example CD4.
8. The method according to any one of claims 1 to 7, wherein said blood sample
is selected
from peripheral, capillary or venous blood samples or subtractions thereof,
such as, for
example, peripheral blood monocytes, blood clots, and dried blood spots.
9. The method according to any of claims 3 to 8, further comprising the step
of concluding on
the immune status of a mammal based on said quantification.

29
10. A diagnostic kit, comprising materials for performing the method according
to any of
claims 1 to 9, optionally with instructions for use.
11. Use of the kit according to claim 10 for performing a method according to
any of claims 1
to 3.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
1
Improved method for epigenetic immune cell counting
The present invention relates to improved methods for epigenetic blood and
immune cell
counting, and respective uses and kits.
Background of the invention
Deviation from the physiological balance of cellular immune system is
indicative of various
diseases and therefore constitutes an important measure for diagnosis and
patient monitoring.
The according measurements are currently best performed by flow cytometry
(FCM), which
provides both accuracy and flexibility with respect to the cell type to be
determined (1).
However and although hematology analyzers used in diagnostic laboratories are
highly
developed and logistic environments are extensively adapted to the required
processes, this
approach is limited in its applicability.
FCM-based cell counting requires fresh, anti-coagulated or well-preserved
blood samples
with intact leukocytes. Even with fresh samples, it is advisable to work
quickly since time-to-
analysis can influence the results with cell deterioration beginning in the
initial hours after
blood draw. Moreover, standardization of FCM remains a significant challenge
due to
biological, technical and operational variation (2,3,4,5) and completely
standardized
measurements have not yet been achieved (6, 7). The most critical challenge,
however, is that
not all medical applications warrant availability of fresh or well-conserved
blood samples and
flow cytometry cannot be applied in these cases.
For example, therapy decisions for HIV infected patients hinge on the
patients' CD4 ' T helper
cell count. At a frequency below 500 T helper cells per microliter,
antiretroviral therapy is
strongly recommended and becomes imperative at levels below 200 cells per
microliter. In
resource-poor countries, an appropriate diagnostic assessment is often
hampered in rural areas
where blood draw and measurement cannot be performed in close succession. As a
result,
treatment is frequently initiated based solely on HIV-related clinical
symptoms, which can
result in suboptimal outcomes (8, 9).
Another example is newborn screening. Guthrie cards from heel pricks are
collected and used
for the detection of severe, curable inborn defects. These cards cannot be
used for flow

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
2
cytometric analysis and T-cell receptor excision circles (TREC) are used for
PID-screening
instead. TREC analysis preferentially detects recent thymic emigrants, the
predominant T-cell
subtype in the periphery of newborns. However, this technology is limited to T-
and, more
recently also B- (KREC), cells but is not fit for differential analysis, such
as CD4 or CD8
subpopulations and also fails to detect other cell types, such as NK cells.
Therefore, TREC
analysis in newborn analysis is exclusively used for initial screening.
Differential diagnosis
and patient monitoring prior to and upon the curative hematopoetic stem cell
transplantation
requires change of technology and is performed by flow cytometry.
In order to overcome diagnostic limitations and the associated technological
switches, an
improved means for immune status assessment would be valuable. It should
robustly provide
relative and absolute cell counts equally allowing the use of fresh, frozen or
paper-spotted
blood. Signals should be digital for each analyzed cell, i.e., indicating
either one positive or
negative value per cell rather than requiring arbitrarily defined thresholds
for "positiveness".
Such new method should also be performed in an automated, operator-independent
manner
and less dependent on the variability of reagents used, such as antibodies.
In a first aspect of the present invention, the above object is solved by a
method for a
quantitative methylation assay for blood immune cells, comprising the steps of
a) providing a defined volume of a sample of human blood comprising diploid
genomic DNA
of blood immune cells to be quantitated;
b) providing an in silico bisulfite-converted recombinant nucleic acid
comprising at least one
demethylation standard gene, and a sequence inversing all CpG dinucleotides to
GpC of said
at least one demethylation standard gene ("standard I");
c) providing a recombinant nucleic acid comprising the demethylated genomic
sequence of
said at least one demethylation standard gene of b), and of said a sequence
inversing all CpG
dinucleotides to GpC of said at least one demethylation standard gene of b)
("calibrator I");
d) providing a recombinant nucleic acid comprising the sequence inversing all
CpG
dinucleotides to GpC of said at least one demethylation standard gene of b)
("spiker I");
e) adding a defined amount of said recombinant nucleic acid of d) to said
sample of a)
("spiking");
f) treating said diploid genomic DNA of the cells to be quantitated of a) and
said recombinant
nucleic acids of c) and d) with bisulfite to convert unmethylated cytosines
into uracil;

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
3
g) amplifying of said nucleic acid molecules of a), b), c), and f) using
suitable primer pairs to
produce amplicons; and
h) identifying the blood immune cells (BIC) per volume of sample based on
analyzing said
amplicons.
In a second aspect of the present invention, the above object is solved by a
method for a
quantitative methylation assay for blood cells, comprising the steps of
a) providing a sample of human blood comprising diploid genomic DNA of blood
cells to be
quantitated;
b) providing an in silico bisulfite-converted recombinant nucleic acid
comprising at least one
demethylation standard gene, and at least one blood cell specific gene
("standard II");
c) providing a recombinant nucleic acid comprising the demethylated genomic
sequence of
said at least one demethylation standard gene of b), and of said at least one
blood cell specific
gene of b) ("calibrator II");
d) treating said diploid genomic DNA of the cells to be quantitated of a) and
said recombinant
nucleic acid of c) with bisulfite to convert unmethylated cytosines into
uracil;
e) amplifying of said nucleic acid molecules of a), b), c), and d) using
suitable primer pairs to
produce amplicons; and
0 identifying the percent of demethylation per all cells (DDC) based on
analyzing said
amplicons.
In a particularly preferred third aspect of the present invention, the above
object is solved by a
method for determining the absolute copy number of methylated genes of an
immune cell
type, comprising
a) Performing the method according to the first aspect of the invention as
above,
b) Performing the method according to the second aspect of the invention as
above, and
c) Multiplying the BIC as identified with the DDC as identified.
The steps of the methods of the present invention can be performed in parallel
or combined
reaction vials, as shown in Figure 2.
The present invention relates to the accurate quantification of methylation
data. This involves
several components and considerations:

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
4
1. An internal standard, e.g. in silico converted plasmids.
2. A (e.g.) GAPDH normaliser in contrast to the methylated variant of a
specific gene.
3. Thus, a comparison of all demethylated copies by the obligatory
demethylated GAPDH
with the specific (but present in the same number of copies) demethylated gene
according to
the quantification with 1.
4. Nevertheless, the above does not allow a truly "absolute" quantification,
since the in silico
converted standard does not correspond to the biological sample (which is
converted only in
the reaction vial.
5. Solving the problem at 4. based on adding and measuring a so-called GNoMs
(Genomic
Normaliser of Methylation), here, all original sequences are equimolarly
included into a
plasmid and then submitted to the overall process (bisulfite treatment and
purification). Since
they are present 1:1 a standard can be identified after the quantification
using the standards in
1 showing the difference between in silico and in situ methylation. Using this
factor, the
methylation value of the measurements can be corrected, which improves the
result
considerably.
6. Using a defined amount of a nucleic acid (plasmid) with a standard gene
having inverted
CG bases, furthermore, any loss of material during the process can be
accounted for, which
further improves the method.
In principle, quantitative real-time PCRs (qPCR) is such an approach
accomplishing the
characteristics when based on highly cell-type specific epigenetic (i.e., DNA
methylation)
markers (14, 15, 16). Technically, when DNA is treated with bisulfite,
unmethylated CpG
dinucleotides are converted to TpGs ("TpG-variant"), whereas methylated CpG-
dinucleotides
remain unaltered ("CpG-variant"). Thus, bisulfite conversion translates
epigenetic marks into
sequence information allowing discrimination between - and quantification of
both - variants.
qPCR assays are non-susceptible to loss of cell integrity in a blood sample,
since its DNA is a
very stable entity. Epigenetic cell quantification can be thus performed on
fresh peripheral
blood, dried blood spots or any other specimens without particular demands on
their state of
preservation. One can assume the existence of two copies of each CpG locus per
cell and that
each locus is exclusively either methylated or demethylated in a single given
cell. In addition,
the essential components required for qPCR are synthetically produced and
standardization is
relatively easy to achieve when compared with the manufacturing of biological
substances
like specific antibodies. However, as of today, the applicability of such
epigenetic approach
has not been demonstrated, probably due to the absence of well-defined cell
type specific

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
biomarkers and a lack of methods to implement definitive and absolute
quantification. .
Definitive quantification requires reference standards directly representing
the biological
substrate (17). Absolute quantification poses difficulties in DNA-based
techniques since the
relation between leukocyte DNA and blood volume is not biologically fixed and
DNA
recovery from blood extraction is semi-quantitative.
Here, the inventors introduce a panel of epigenetic immune cell specific qPCR-
based assays
for quantification of the major leukocyte populations in human blood samples.
The assays are
based on DNA methylation marks specific for overall T cells, CD4 ' T cells,
CD8 ' T cellsõ B-
cells and NK cells. The cell number per microliter of blood constitutes the
standard of care
when fresh blood samples are available e.g., for the determination of CD4+T
cells in HIV
striken patients. Therefore, the inventors propose a novel system for
definitive and absolute
counting of immune cells based on their cell type-specific epigenetic signals.
The assay
concept and the individual biomarkers were analyzed for their equivalence to
the gold
standard FCM technology. However, when the volume of a given blood is not
exactly
defined, as is the case for dried blood samples, relative measurement is more
accurate and
should be employed. Hence and in order to achieve a wide measurement spectrum,
here the
inventors tested various diagnostic applications, aiming at the degree of
agreement of absolute
quantification by analysis of healthy donors and a cohort of HIV-positive
patients, but the
inventors also determined the diagnostic quality by identifying PID patients
within a cohort of
healthy newborns cohort from dried blood spots.
In a preferred embodiment of the method(s) according to the present invention,
the method is
integrated, and comprises
a) providing a defined volume of a sample of human blood comprising diploid
genomic DNA
of blood cells to be quantitated;
b) providing an in silico bisulfite-converted recombinant nucleic acid
comprising a
demethylation standard gene, a sequence inversing all CpG dinucleotides to GpC
of said
demethylation standard gene, and a blood cell specific gene;
c) providing a recombinant nucleic acid comprising the demethylated genomic
sequence of
said demethylation standard gene of b), a sequence inversing all CpG
dinucleotides to GpC of
said demethylation standard gene, and said blood cell specific gene of b);
d) providing a recombinant nucleic acid comprising the sequence inversing all
CpG
dinucleotides to GpC of said at least one demethylation standard gene of b);

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
6
e) adding a defined amount of said recombinant nucleic acid of d) to said
sample of a)
("spiking");
f) treating said diploid genomic DNA of the cells to be quantitated of a) and
said recombinant
nucleic acids of c) and d) with bisulfite to convert unmethylated cytosines
into uracil;
g) amplifying of said nucleic acid molecules of a), b), c), and f) using
suitable primer pairs to
produce amplicons; and
h) identifying the amount of blood cells per volume of sample based on
analyzing said
amplicons.
Preferably, the nucleic acids are plasmids, e.g. linearized plasmids, such as
bacterial plasmids,
e.g. pUC.
In this aspect of the method, the amplification is normalized using a first in
silico bisulfite
converted nucleic acid (plasmid), comprising a demethylation standard gene
(e.g. GAPDH),
an "artificial sequence" (the sequence inversing all CpG dinucleotides to
GpC), as well as a
blood cell specific gene (a "specific gene"). All three elements are equally
present
(equimolar) on said nucleic acid, and are in silico bisulfite converted.
Therefore, the
normalization curve and the corresponding calibration curves can be directly
compared with
the sample, and the relative cell count can be determined from the ratio of
blood cell specific
gene to demethylation standard gene. Nevertheless, the nucleic acid does not
correspond to
the "real" sequence, since each C is replaced by a T. A serial dilution and
determination of
each concentration with all genes as mentioned generated the calibration curve
for the assay.
In order to improve the accuracy of the approach, a second nucleic acid
(plasmid) is used
comprising the demethylation standard gene (e.g. GAPDH), the "artificial
sequence" (the
sequence inversing all CpG dinucleotides to GpC), and the blood cell specific
gene (a
"specific gene"). Nevertheless, these sequences are NOT in silico bisulfite
converted, and
correspond to the genomic sequences (in as far as the have a genomic
counterpart, see below)
¨ and thus can only be used for measuring the amplification (e.g. qPCR)
efficiency.
The reason for the second standard is two-fold. A) For a definitive
quantification a standard is
required that is identical as in the biological sample to be analyzed (this is
also a regulatory
requirement). In the first nucleic acid, nevertheless, a double stranded AT-
rich sequence is
compared with a single-stranded U-rich sequence. Only the "true" bisulfite
conversion of the

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
7
double stranded nucleic acid allows for this definitive comparison. Then, the
quotient of
bisulfite conversion of blood cell specific gene to demethylation standard
gene, normalized
using the first nucleic acid, gives a factor of the efficiency. The same holds
true for a quotient
based on the division of the bisulfite conversion of the sequence inversing
all CpG
dinucleotides to GpC by the bisulfite conversion of the demethylation standard
gene.
Preferably, the "artificial sequence" (the sequence inversing all CpG
dinucleotides to GpC) is
a random sequence comprising C and CpG sequences (for bisulfite conversion)
that does not
occur in the human genome. In one embodiment, the artificial sequence is the
exact sequence
of the part of GAPDH that is amplified (amplicon) wherein the CpG sequences
are inverted
into GpC sequences. The "artificial sequence" is found on all three nucleic
acids as described
above, namely on the first one (in silico bisulfite converted), the second one
(for bisulfite
conversion), and - as the only analyzed sequence - on the third nucleic acid
(in silico bisulfite
converted).
The third nucleic acid is given in a defined amount into a defined amount of
blood, and is then
analyzed (e.g. purification, bisulfite treatment, second purification,
desulfonation, specific
amplification). Then, a normalization is performed against the first nucleic
acid (how many
copies were measured and given into the reaction), the efficiency is
determined using a
comparison with the second nucleic acid, and the (residual) copy number is
determined using
the third nucleic acid. Any losses are compared with a loss of genomic DNA
that was
subjected to the same procedure. The overall process allows for a precise
definitive and
absolute quantification of said DNA, and through this the cells in a blood
sample, such as, for
example, whole blood.
In one embodiment, the invention relates to an artificial sequence that is the
exact sequence of
the part of GAPDH that is amplified (amplicon) wherein the CpG sequences are
inverted into
GpC sequences as a tool when performing the method(s) of the present
invention.
The composition of the cellular immune system holds valuable diagnostic
information for
various diseases. The standard technology for quantitative immune cell
monitoring is flow
cytometry. However, this method is limited to blood samples in which cell-
integrity is
retained. In clinical routine, this effectively restricts analysis to fresh
blood samples as
analytical substrate.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
8
In order to widen the margin of use of diagnostic immune monitoring, the
inventors
implemented epigenetic qPCR systems for quantification of the major leukocyte
populations.
Upon determining immune cell type specific methylation marks, whole blood from
25 healthy
donors, 97 HIV patients and 325 Guthrie cards from newborns including 25 cards
from
patients with primary immunodeficiencies (PID) were analyzed. Methodological
concordance
between flow cytometric and epigenetic data for B-, NK-, total T cells, T
helper cells and
cytotoxic T cells was determined and the ability of this new technique to
identify quantitative
immune cell deficiencies was challenged.
Data show that quantification via epigenetic qPCR assays and flow cytometry
perform
equivalently in healthy subjects and HIV patients according to Bland-Altman
testing.
Epigenetic quantification is applicable for relative and absolute frequencies
of leukocyte
subsets in fresh and frozen blood samples. In contrast to flow cytometry,
immune cell analysis
of Guthrie cards accurately identifies cases PID in newborns. Epigenetic
quantification of
immune cell populations performs with high equivalence to standard flow
cytometry offering
a wider range of possible applications, including analysis of dried blood
spots possibly laying
a path to blood counting of patients in remote areas or from newborns.
Preferred is the method according to the present invention, wherein said blood
immune cell is
selected from a leukocyte, a T-lymphocyte, a granulocyte, a monocyte, a B-
lymphocyte
and/or an NK-cell.
Preferred is the method according to the present invention, wherein said
recombinant nucleic
acid molecule is selected from a plasmid, a yeast artificial chromosome (YAC),
human
artificial chromosome (HAC), PI-derived artificial chromosome (PAC), a
bacterial artificial
chromosome (BAC), and a PCR-product.
Preferred is the method according to the present invention, wherein said
demethylation
standard gene is selected from a gene expressed in all cells to be detected,
such as, for
example, a housekeeping gene, such as, for example, GAPDH.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
9
Preferred is the method according to the present invention, wherein said blood
cell specific
gene is selected from a gene expressed in all blood cells to be detected, such
as, for example
CD4.
Preferred is the method according to the present invention, wherein said blood
sample is
selected from peripheral, capillary or venous blood samples or subtractions
thereof, such as,
for example, peripheral blood monocytes, blood clots, and dried blood spots.
Preferred is the method according to the present invention, further comprising
the step of
concluding on the immune status of a mammal based on said quantification.
Another aspect of the invention relates to a diagnostic kit, comprising
materials for
performing the method according to the present invention, optionally with
instructions for
use. Preferred materials are the nucleic acid molecules, and/or a bisulphite
reagent.
Another aspect of the invention relates to the use of the kit according to the
invention for
performing a method according to the invention.
Current immune cell monitoring requires fresh or well-conserved blood samples.
Here, the
inventors present an alternative technology for differential blood cell
counting that allows
calculation of relative and absolute cell numbers in fresh, formalin-fixed,
frozen or dried
blood samples.
Epigenetic qPCRs emerged as a promising tool without a need for intact cells
or high
standards of preservation. Data reporting the demethylation of the FOXP3 TSDR
in
regulatory T cells (14) showed the feasibility of such epigenetic cell
counting.
In this invention, still open questions were addressed regarding a) the
availability of feasible
epigenetic markers for several important cell types, b) the normalization
between various cell-
type specific epigenetic qPCR systems, c) approaches to provide definitive
quantification (17)
for the epigenetic signals d) the quantification relative to a given blood
volume and e)
methodological agreement with the FCM gold standard technology to detect the
according
immune cell types.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
Ideal DNA-methylation markers for cell-type identification are discriminative
between the
cell type of interest (near 0% methylation) and all other cell types (near
100% methylation).
Here, markers for immune cell counting were investigated, initially applying
the not fully
quantitative method of bisulfite sequencing at putative discriminatory loci.
These data show
loci for all tested immune cell types tentatively fulfilling the criterion for
ideal markers.
Discriminatory CpG-dinucleotides were selected for qPCR assay development and
tested on
methylated and demethylated template variants. Efficient and quantitative
amplification of
target DNA was achieved without detecting background from non-target
templates. qPCR
assay performance was robust with low deviation in fresh or frozen blood.
For simultaneous testing of various different cell types in a heterogeneous
sample, the use of
locus-specific methylated (i.e., CpG) and demethylated (TpG) qPCRs is not
optimal.
Amplification efficiency varies for each qPCR system e.g., due to differing
CpG-density, so
that there is no biologically or technically stable parameter for all
measurements (20). An
invariably demethylated regulatory stretch of the housekeeping GAPDH (21) gene
was used
instead as a fixed denominator. In this way, each cell-type specific locus is
counted relative to
the number of loci demethylated in GAPDH, i.e., supposedly in all nucleated
cells. Testing
purified cell types at their specific epigenetic loci, however, showed that
quantification with
GAPDH as denominator deviates from quantification obtained with the locus-
specifically
methylated amplification systems. These deviations are dependent on the
individual locus and
amplification systems (22, 23) suggesting that normalization with a standard
curve from in
silico converted plasmid does not fully compensate for amplification
differences in de facto
bisulfite-converted DNA. Plasmid DNA is double-stranded and contains
approximately 40%
GC (24) outside of the templates for the epigenetic amplification. In
contrast, bisulfite
converted DNA is predominantly single-stranded with a significantly reduced GC
content
after amplification. Moreover, the latter has been exposed to harsh chemical
treatment causing
fragmentation which is not physically represented by in silico conversion of
demethylated
cytosine to thymine.
For compensation of this effect, a single plasmid molecule ("calibrator")
containing the
original demethylated genomic sequences of GAPDH and all immune cell-type
specific
marker genes was processed along with the biological samples. This calibrator
provides for
relative and equimolar quantification of bisulfite converted DNA via different
qPCR systems.
It does not replace the standard plasmid, however, since precise independent
quantification

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
11
along bisulfite conversion is challenging. Hence, in silico bisulfite-
converted plasmid
("standard") continues to be used for copy number determination. The
calibrator plasmid
provided highly reproducible measurements throughout various experiments and
compensates
for the observed efficiency differences between GAPDH and the locus specific
assays. Taken
together, the parallel use of standard and calibrator plasmids renders the
epigenetic qPCR into
a method for definitive quantification (17) of demethylation in the respective
loci relative to
all (demethylated) GAPDH copies.
The data in Figure 1 and Table 1 suggest that the analysis of differentially
methylated loci is a
powerful surrogate for detecting and quantifying defined immune cell types. In
addition of
cell type specificity, however, a direct proportional relation of cell type
and DNA copy
number needs to be experimentally established if cell counting by this means
is intended.
Such direct linear association may be impugned by residual DNA in
thrombocytes,
reticulocytes or by shifts between DNA-copies and cell numbers. Thus, cell
quantification by
epigenetic qPCR was orthogonally compared with FCM analysis. When using the
definitive
relative quantification introduced in this work, data for individual cell
types and markers
differed mildly with systemic biases ranging from -6% to +11%. Overall,
however, high
methodological agreement was observed between both technologies as whole and
the selected
individual markers.
With respect to routine clinical applications, relative cell type
quantification is accepted by
WHO in HIV-treatment guidelines, but medical reality demands cell counting per
volume (25,
26). For epigenetic immune cell typing this poses a problem, since DNA
recovery is not fully
quantitative and a relation between DNA amount and blood volume is not
biologically
determined. Spiking a defined concentration of the inverse GAPDH variant (GAP)
into
blood samples, however, allowed an approximated inference to the original DNA
content
prior to DNA extraction. Whereas different efficiencies of genomic DNA and
plasmid DNA
have been described (27), such difference might be significantly reduced after
bisulfite
treatment and resulting fragmentation. For evaluation of the spiking concept,
epigenetic
immune cell counting was paralleled with flow cytometry data from the same
blood samples.
With respect to bias and limits of agreements (28) the presented data show
homogenous error
distribution and smaller deviations than previously published methodological
comparisons
among different antibody-based methods (29). Hence, the inventors' data
indicate that

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
12
leukocyte subsets can be detected reliably by epigenetic immune cell counting
and are almost
comparable to FCM data.
An intrinsic challenge for quantitative diagnostic markers is their reliable
performance outside
the normal physiological range and from frugally conserved samples. For immune
cell
quantification, PIDs constitute such situation. Patients suffer from severe
immune
deficiencies, and quantitative cell counting via FCM is not feasible from
dried blood cards for
newborn screening.
In such case, comparison with the diagnostic performance of the gold standard
method may
best inform about the diagnostic power of the novel method. As direct method
comparison is
not feasible due to differing measurement parameters between TREC counting
compared to
cell quantification by FCM and/or epigenetic qPCR, such outcome comparison
appears
indispensable. Epigenetic immune cell counting reliably identifies all PID
patients from dried
blood spots, including XLA patients. In addition, it provides information on
NK cell levels ¨
an important hint for the underlying genetic defect, especially for ADA-SCID,
since in these
delayed onset form of SCID NK cells are - contrary to other forms of SCID ¨
missing.
Taken together, the present invention shows that epigenetic cell counting
provides precise and
accurate means for immune monitoring, regardless if measured as continuous
parameter in a
percentage or absolute cell count or as categorical parameter. When detecting
lower method
agreement, as observed between the epigenetic and FCM markers for monocytes,
this
suggests that heterogeneity within a given cell population or ambiguous marker
specificity
influence agreement.
Altogether, this invention underlines the applicability of epigenetic immune
monitoring as a
valuable diagnostic way to immune diagnostics by providing a highly robust
platform capable
for analyzing samples from minute amounts of frugally conserved blood.
The present invention will now be explained further in the following examples
and figures,
nevertheless, without being limited thereto. For the purposes of the present
invention, all
references as cited herein are incorporated by reference in their entireties.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
13
Figure 1 shows the DNA methylation profile of marker genes in purified immune
cell
populations. The matrix shows cell type-specific DNA methylation patterns of
seven marker
genes and of the reference gene GAPDH. In the matrix, immune cell types are
arranged in
columns as indicated at the x-axis. Genes and corresponding amplicons (Amp)
are indicated
at the y-axis. Genes are separated by red lines with each row representing a
single CpG site.
Measured CpG methylation levels are color coded according to the color scale
ranging from
yellow (0% methylation) to blue (100% methylation).
Figure 2 shows a schematic overview over the different quantification
approaches for
epigenetic cell counting. For all approaches, the inventors assumed a
simplified 2-allels-to-1-
cell relation. Each analyzed gene in this invention was autosomal (i.e.,
diploid), and has been
shown to be demethylated in one specific cell type (in this scheme: the CD4
locus in CD4 ' T
helper cells) whereas completely methylated in all other blood cell types. A)
sketches the
process for locus-specific relative percentage quantification. In blood
samples with an
unknown number (#) of demethylated and methylated diploid genomic DNA copies,
a
bisulfite conversion transfers the epigenetic methylation status into the
primary sequence by
exclusively converting demethylated Cytosins into Uracils. In CpG positions,
methylated and
demethylated cytosins occur depending on specific gene regulation. Converted
Uracil-DNA is
CpG-methylation status specific amplified by qPCR, whereby Uracil basepairs
with adenosine
resulting in an amplificate containing TpG dinucleotides at originally
demethylated regions.
qPCRs then allow counting of copy numbers as based on the calculation of
serially diluted in
silico converted plasmids by a linear interpolation (f-1) of the amplification
results (f).
Relative percentage methylation at the genomic locus is calculated by the
interpolated copy
number of originally demethylated copies at this locus divided by all copies
at this locus, i.e.,
the methylated and demethylated ones. Conversion in the biological sample
perturbs the
integrity of the genomic DNA, whereas the plasmid represents the amplification
product and
not the substrate. The resulting difference in amplification efficiency is
given by an unknown
"conversion factor, (CF)". It is considered negligible when comparing
amplification of two
highly homologous sequences with few methylation-status dependent SNPs. For
universal
relative percentage qPCR (B), the same principles for epigenetic
quantification are employed
with regard to using an in silico converted plasmid standard, its
interpolation and a cell type
specific demethylated gene locus. Instead of the assessment of the cell
specifically
demethylated locus, the universally demethylated GAPDH locus representative
for all cells is
amplified. Using this as quantification reference all specific loci can be
normalized to the

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
14
overall genomic copy count. CF cannot be assumed to be similar in this case,
as no homology
between the different sequences is assumed. Relative demethylation per all
cells is therefore
disturbed by the presence of differing CFs. Compensating the influence of
different
conversion efficiencies, a calibrator plasmid is introduced as indicated in
C). It contains
equimolar genomics sequences of all relevant cell-specific loci and GAPDH.
Interpolation of
the amplification provides copy numbers for interference of differing
conversion-specific
efficiencies. The ratio of the differing copy numbers provides an efficiency
factor (EF) that
can be used to eliminate conversion related differences between standards and
samples.
Incorporating EF into (B) provides for definitive copy number quantification.
D) For counting
cells per volume of blood, a defined volume of blood is supplemented with
known copy
number of plasmid containing a synthetic, not natural DNA sequence (GAP-GC).
Relative
quantification using the in silico converted plasmid and calculation of EF
operates as
indicated above. Interpolating the starting amount of GAP-GC allows monitoring
of DNA
preparation, conversion and qPCR provides a good estimator for process
efficacy. Calculation
of the starting amount of blood cells therewith becomes possible.
Figure 3 shows the comparison of immune cell quantification by flow cytometry
and
epigenetic qPCR in blood from 25 healthy donors. Immune cells as measured via
flow
cytometry (y-axis) were scatter plotted over corresponding values determined
via immune cell
type specific epigenetic qPCR analysis (x-axis). A) shows relative immune cell
counting
where values are given in percent among total leukocytes. Linear Pearson
correlation
coefficients were r=0.95. B) displays absolute immune cell counting where
values are
expressed as cell number per 1 of blood featuring a correlation of r=0.95.
The red line
represents the regression line computed from all data points, the black line
indicates the
bisectrix. Symbols in the right-hand legend illustrate the different
individual immune cell
populations.
Figure 4 shows the method comparison between flow cytometry and epigenetic
qPCR
analysis of T-cell subsets in a HIV cohort. A) illustrates the comparison of
relative immune
cell counts (as expressed in % related to total nucleated cells). The large
graph shows a scatter
plot of three T cell populations analyzed via epigenetic qPCR analysis (x-
axis) and flow
cytometry (y-axis). The lines in black and red represent the bisectrix and the
regression line,
respectively. Linear correlation coefficient according to Pearson was r= 0.982
(p<0.0001).
The small graphs display a Bland-Altman analysis where the mean cell count (in
%) as

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
averaged between each epigenetic and cytometric measurement (x-axis) was
plotted over their
(relative) difference (y-axis). In each Bland-Altman plot, the upper and lower
red lines reflect
the limits of concordance and the central red line illustrates the systematic
bias. Above and
below each red line, the 95% confidence interval is shown as dotted grey
lines. Upper Bland-
Altman panel: Total T cells; bias: 6.43%; lower limit of agreement: -9.15%;
upper limit of
agreement: 22.02%. Middle panel: Cytotoxic T cells; bias: 11.23%; lower limit
of agreement:
-15.36%; upper limit of agreement: 37.83%. Bottom right: Helper T cells; bias:
-6.04%; lower
limit of agreement: -41.34%; upper limit of agreement: 29.25%. B) shows the
comparison of
absolute immune cell counts (as expressed in cells per 1 blood). Left side:
Scatter plot
analysis; Pearson r=0.955 (p<0.0001). Right side: Bland-Altman analysis: Upper
panel: Total
T cells; bias: -4.76%; lower limit of agreement: -39.62%; upper limit of
agreement: 30.09%.
Middle panel: Cytotoxic T cells; bias: 0.03%; lower limit of agreement: -
35.78%; upper limit
of agreement: 35.83%. Bottom right: Helper T cells; bias: -17.61%; lower limit
of agreement:
-59.68%; upper limit of agreement: 24.46%.
Figure 5 shows the analysis of control Guthrie cards and PID cases with
epigenetic markers
for CD3+ T cells, CD19+ B cells, CD56+ NK cells (as well as CD4 and CD8 T
cells below).
Healthy controls are given in grey dots and 99% (blue) and 99.9% (red)
confidence intervals
of a bivariate normal distribution are estimated from log transformed copy
numbers.
Measurement data of the test cohort are given as numbers and color-coded
according to
phenotype.
Examples
Abbreviations: Amp, amplicon; qPCR, quantitative real time polymerase chain
reaction;
FCM, flow cytometry; HSCT, hematopoietic stem cell transplantation. RDis,
locus-specific
relative demethylation; RD, universal relative demethylation; DD, universal
definitive
demethylation; LC, leukocyte count; CF, conversion factor.
Leukocyte populations. Peripheral blood samples were obtained from healthy
donors and
fractionated into CD15 ' granulocytes, CD14 ' monocytes, CD56 ' natural killer
cells, CD19 '
B-lymphocytes, CD3 'CD4 ' T-helper cells and CD3 'CD8 ' cytotoxic T cells by
high-speed
fluorescence activated cell sorting as described previously (16). Purities of
sorted cells were
>97% as determined by flow cytometry and viability was >99%.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
16
Peripheral whole blood samples. EDTA-anticoagulated peripheral blood samples
were
collected from 25 healthy subjects from each one blood draw, 97 HIV ' patients
under
treatment (each one blood draw) in a German outpatient clinic and 26 patients
with (acute
myeloid) leukemia from San Raffaele University Hospital receiving
hematopoietic stem cell
transplantation. From the latter cohort 92 blood draws from conditioning phase
to 180 days
post transplantation were made. All samples were subjected in parallel to
epigenetic qPCR
analysis and to standard flow cytometry analysis for immune cell
quantification (see below)
without need for additional venipuncture according to Medical device act.
Ethical consent was
given at the according institutions. For epigenetic analysis, all data were
blinded to
experimenters. For diagnostic FACS analysis, samples were not blinded.
DNA preparation. For sequencing and qPCR analysis of purified immune cells,
genomic
DNA was isolated using DNeasy tissue kit (Qiagen) according to the
manufacturer's
instructions. In all other applications, blood samples were forwarded to a one-
tube lysis and
bisulfite conversion without preceding DNA preparation.
Bisulfite conversion. For conversion of purified genomic or plasmid DNA, the
EpiTect Fast
Bisulfite Conversion Kit (Qiagen) was used following the manufactures
protocol. For direct
bisulfite conversion of whole blood, 20 1 of EDTA anti-coagulated blood (or
calibrator
plasmid) was mixed with 16 1 lysis buffer, 3 1proteinase K (Qiagen) and, where
appropriate,
1 1 of GAP [Gcl spiker plasmid yielding 20,000 copies/ 1 blood followed by
incubation at
59 C for 10 minutes. For conversion, 90 1 ammonium bisulfite (68-72%, pH 4.8-
5.3, Chemos
AG) and 341 tetrahydrofurfuryl alcohol (Sigma-Aldrich) were added. Conversion
and
purification of converted DNA was carried out according to the "EpiTect Fast
Bisulfite
Conversion Kit" protocol.
Bisulfite sequencing. PCR-amplification was performed in a final volume of 25
1 containing
lx PCR Buffer, 1U Taq DNA polymerase (Qiagen), 200 M dNTPs, 12.5pmo1 each of
forward and reverse primers, and approx. lOng of bisulfite-converted genomic
DNA at 95 C
for 15 minutes and 40 cycles of 95 C for 1 minute, 55 C for 45 seconds and 72
C for 1
minute and a final extension step of 10 minutes at 72 C. PCR products were
purified using
ExoSAP-IT (USB Corp.) and sequenced applying one PCR primer applying ABI Big
Dye

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
17
Terminator v1.1-chemistry (Applied Biosystems) followed by capillary
electrophoresis on an
ABI 3100 genetic analyzer. AB1 files were interpreted using ESME (18).
Epigenetic qPCR analysis. Experiments were performed in a final volume of 10 1
using
Roche LightCycler 480 Probes Master chemistry containing 5Ong lambda-phage DNA
(New
England Biolabs) and up to 100 ng converted DNA template or an adequate amount
of
plasmid. Standard concentration for each primer was at 1.5 M, except for
genomic spiker
plasmid (0.75 M). CD4 T cell TpG assay (4.5 M forward; 3 M reverse primer).
Standard
probe concentration was at 0.25 ILIM except for CD4 ' T cells, CD8+ T cells,
NK cells and
spiker plasmid (each 0.125 M probe for TpG-specific systems). The thermal
profile was
95 C for 10 minutes followed by 50 cycles at 95 C for 15 seconds and 61 C for
1 minute.
Plasmids. Two bisulfite-converted sequences corresponding to either the
methylated or the
demethylated marker regions were designed in silico, synthesized and inserted
into plasmid
pUC57 (Genscript Inc.) and used as positive control for assay establishment
and as
quantification standard for qPCR experiments. Standard plasmids harbour all
assay target
sequences (as TpG- or CpG-variants) and are intra-molecularly linked providing
for
equimolarity of all assay targets. Plasmids were spectrophotometrically
quantified, linearized
by Sca I and serially diluted in lOng/ 1 of lambda-phage DNA (New England
Biolabs) to
obtain quantification standards with 31250, 6250, 1250, 250, 50 or 30 copies
per reaction. For
qPCR normalization, a single calibrator plasmid was generated harbouring all
assay target
sequences equimolarly in the genomic unconverted demethylated version. For
leukocyte
quantification per 1 blood, a spike-in plasmid was designed and generated
carrying an
unconverted artificial GAPDH gene region, which is exactly equivalent to the
target of the
GAPDH-specific qPCR assay but has all CpG dinucleotides inverted to GpCs
(GAP).
Oligonucleotides. Forward (fp), reverse (rp) primers and hydrolysis probes (p)
(Metabion
AG) are indicated by their chromosomal positions with respect to the human
genome
assembly GRCh38.p5, Release 84 (March 2016). Oligonucleotides for bisulfite
sequencing:
AMP1255: fp: 12:6790192-214, rp: 12:6790582-603; AMP1730: fp: 9:128149251-72,
rp:
9:128149589-609; AMP2000: fp: 12:6790724-46, rp: 12:6791160-80; AMP2001: fp:
12:6791141-62, rp 12:6791535-60; AMP2007: fp: 2:86821232-54, rp 2:86821674-95;
AMP2178: fp: 6:161375641-62, rp 6:161376086-108; AMP2249: fp: 11:68371460-81,
rp:
11:68371926-47; AMP2674: fp: 16:88653882-902, rp: 16:88654299-88654320.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
18
Oligonucleotides for qPCR analysis: CD4: TpG: fp: 12:6790871-98, rp:
12:6791046-73, p:
12:6790998-1019; CpG: fp: 12:6790871-900, rp: 12:6791046-72, p: 12:6790997-
1020.
CD8B: TpG: fp: 2:86821374-1400, rp: 2:86821476-93, p: 2:86821425-52; CpG: fp:
2:86821372-1401, rp: 2:86821463-83, p: 2:86821425-55. LPR5: TpG: fp:
11:68371608-28,
rp: 11:68371721-45, p: 11:68371666-84; CpG: fp: 11:68371611-35, rp:
11:68371720-48, p:
11:68371662-86. MVD: TpG: fp: 16:88654112-36, rp: 16:88654173-90, p:
16:88654136-55;
CpG: fp: 16:88654111-36, rp: 16:88654172-89, p: 16:88654136-58. PARK2: TpG:
fp:
6:161375730-55, rp: 6:161375851-66, p: 6:161375804-25; CpG: fp: 6:161375784-
807, rp:
6:161375851-70, p: 6:161375805-830. LCN2: TpG: fp: 9:128149258-78, rp:
9:128149353-
75, p: 9:128149289-309; CpG: fp: 9:128149257-77, rp: 9:128149353-76, p:
9:128149287-
309. Oligonucleotides of the CD3 ' T cell and GAPDH-specific amplicons and
qPCR-systems
have been published previously (15).
Flow cytometric characterization of whole blood samples - To compare results
of the
epigentic analyses to standard flow cytometry, the absolute number of CD45 '
leukocytes was
determined after lysis of erythrocytes by a MACSQuant cytometer (Milteny
Biotec, Bergisch
Gladbach). In addition frequencies and absolute counts of CD15 ' granulocytes,
CD14 '
monocytes, CD19 B-cells, CD56' NK cells, total CD3 'T cells and CD4 ' and CD8'
subsets
were calculated as previously described (14,32).
Statistical analysis - CP (õcrossing point") of aggregated triplicate
measurements was
computed by the second-derivative maximum method applying the LC480 software
(Roche)
to yield copy numbers ("plasmid units") by interpolation (f-1) of
amplification (f) from
calibration curves generated with dilutions of plasmid-based standards. Method
comparison
between flow cytometric and qPCR based measuring technique was done as
follows:
Bivariate data from the two methods were drawn in a scatterplot. Linear
Regression was
performed testing a) for a slope different from 1 and b) an intercept
different from 0. Bland-
Altman plots were inspected analyzing bias and precision statistics (28).
Acceptable precision
was regarded as average deviation from the bias in percent, reflecting the in
house limit on the
coefficient of variation for intra assay performance, i.e., 0.2. This
translates into acceptable
limits of agreement of 0.4. The inventors report the estimated bias, the
precision statistic and
the respective 95% confidence intervals. For correlation, Pearson product-
moment
correlations were used. Rater agreement was evaluated using Cohens-Kappa
coefficient (19).
All p-values are two-sided. Statistics software R 3.3.0 was employed.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
19
Cell type-specific bisulfite-conversion. Methylation-dependent conversion of
CpG-
dinucleotides was analyzed by bisulfite sequencing (18) aiming at marker
identification for
immune cell populations sorted from human peripheral blood. Candidate loci
were selected
from the literature or from a genome-wide discovery experiment. As a likely
marker for CD4 '
T helper cells, the inventors designed three amplicons (Amp) for bisulfite
sequence analysis
covering regulatory elements within the 5' region of the first intron (Amps
1255, 2000 and
2001) in the CD4 gene. Unmethylated CpG-sites are detected as TpG residues
after bisulfite-
conversion and amplification exclusively in the target cells, i.e., CD3 'CD4 T
lymphocytes.
The same CpGs were inert to bisulfite-conversion in control cell types,
including CD56'
natural killer (NK) cells, CD3 'CD8' T lymphocytes, CD14 ' Monocytes, CD19' B-
lymphocytes and CD15 ' Granulocytes (Figure 1). The inventors investigated the
CD8B gene
as a potentially apt epigenetic mark for CD8' cytotoxic T cells by designing
amplicons
targeting regulatory elements within its third intron (Amp 2007). Here,
bisulfite-mediated
conversion of CpGs was observed exclusively in CD3 'T CD8' (target) cells
whilst CpGs
were inert to conversion in control cell types. Similar to data for CD8' and
CD4' T cells, the
inventors identified epigenetic marks, each uniquely demethylated in the
target cell type and
fully methylated in the corresponding control leukocyte populations. Amplicons
corresponding to genes LRP5 (Amp2249) and MVD (Amp2674), served as epigenetic
markers for B cells and NK cells, respectively. The DNA methylation profile of
the intergenic
CD3G and CD3D region (Amps 1405, 1406 and 1408), which constitutes a marker
for CD3 '
T cells and the methylation profile of GAPDH (Amp 1570) were published
previously (15).
Locus-specific relative qPCR measurements. Targeting the differentially
methylated CpG
positions described above, quantitative PCR assay systems were designed as
described in the
method section. The qPCR systems were characterized on in silico bisulfite
converted
template DNA cloned into plasmids (Figure 2A, right panel). For the TpG
template
(mimicking demethylated CpGs in genomic DNA) a universal plasmid carried the
target
regions for all assay and an artificial GAP[Gcl sequence in equimolar
stoichiometry (universal
TpG-plasmid), whereas the "CpG-plasmids" (mimicking methylated CpGs in genomic
DNA)
were designed for each amplicon individually. High technical specificity was
observed with
no cross-reactivity in the mutually antithetic templates (Table 1, "Plasmid-
based controls").
The original copy number of the gene sequences in blood samples was estimated
by relating
the PCR signals from the according amplification (f) to an amplification (f)
of the serially

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
diluted plasmids (Figure 2A). Biological assay specificity was tested on
purified immune cell
populations. High and low copy numbers were observed for target cell types in
the TpG- and
CpG-systems, respectively. Conversely, for control cell types low copy numbers
were found
in the TpG- and high numbers in the CpG-system. Relative demethylation at the
respective
gene loci (RDis) ranged from 89.9 to 100% in target cell types and from 0.0%
to 3% in
controls (Table 1). Exceptions were observed for purified CD4 ' T cells
showing 8.9%
demethylation at the CD8B locus and vice versa (i.e., 9.6% CD4 demethylation
in CD8 T
cells).
Universal and definitive quantification. To provide a joint basis of
quantification for all
cells, the demethylated GAPDH-specific amplification was analyzed together
with the cell-
specific TpG-systems described above (Figure 2B). The universal TpG-plasmid
served as
amplification standard. With this, universal relative demethylation (RD) in
samples was
determined by relating sample amplification f to standard amplification (f)
for each marker
and GAPDH. The inventors' data show that RD u does not always match with
the
corresponding locus-specific demethylation (RDis). To compensate for this
intrinsic
systematic shift a "calibrator plasmid" was adopted harboring all assay
targets in equimolar
amounts and in their unconverted (i.e., demethylated) state. Efficiency
differences between
the individual qPCR systems that remained after standard plasmid based
normalization were
estimated based on the unconverted plasmid and yielded the qPCR efficiency
factor (EF). The
mean EFs between each cell type specific assay and GAPDH were determined in
approx. 25
experiments and ranged between 0.53 for CD4 and 1.17 for CD3x and y (Table 1.
EF).
Application of EF on the universal relative demethylation (RD) then allows for
universal
definitive demethylation quantification (DDu; Figure 2c).
Absolute quantification was established by introducing a "spike-in plasmid"
harboring an
artificial GAPDH sequence inversing all CpG dinucleotides to GpC (GAP) and an
according qPCR assay. Substrate specificity of the GAP-specific qPCR assay was
confirmed on bisulfite converted DNA from whole blood with and without spiker
plasmid
where no cross reactivity with the endogenous GAPDH gene was detected. In
contrast, when
testing the GAPDH-specific qPCR assay on the spike-in plasmid template
harboring an
GAP[Gcl sequence no amplification signal was detected, too demonstrating a
high substrate
specificity, which is indispensable for an absolute quantification. For an
immune cell
counting, the spike-in plasmid was added to blood samples yielding a defined
concentration

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
21
per given initial sample volume (Figure 2D). In addition, the artificial
GAP[Gcl sequence was
included on the in silico bisulfite converted plasmid standard and the
calibrator plasmid
yielding the respective equimolarity factor (EF) of 0.87 (GAP[Gcl; 0.83/0.92)
used for
correction.
Comparative immune cell counting by flow cytometry and epigenetic qPCR. Blood
samples from 25 adult healthy donors were subjected to standard flow cytometry
(FCM) and
epigenetic qPCR for universal quantification of CD15 ' neutrophils, CD14 '
monocytes,
CD19 ' B-cells, CD56 ' NK cells, CD3 ', CD4 ' and CD8 ' T-cells. Data from
both methods
were plotted against each other either as relative (Figure 3A) or absolute
counts (Figure 3B).
Scatter plots indicate for a high level of congruence between both methods
with a Pearson
correlation coefficient r of 0.95 (p<0.0001) for both relative and absolute
quantification of
leukocyte populations. The regression line comparing FCM and epigenetic qPCR
for relative
quantification did not significantly deviate from the bisecting line. However,
a small but
significantly different slope was observed for absolute quantification
indicating a proportional
systematic bias.
To test the inventors' new approach in a real clinical setting the inventors
measured blood
samples from 97 HIV subjects with respect to quantify CD3 ', CD4 ' and CD8 '
cell counts by
standard FCM and epigenetic counting. In this invention, correlation analyses
yielded Pearson
r correlation coefficients ranging from 0.91 to 0.98 (p<0.0001) for relative
and absolute
quantification (Figure 4). Absolute quantification was based on the spiking of
the GAP[Gcl
plasmid into the blood samples to determine the overall leukocyte count
applying the GAP[Gcl
specific qPCR assay system. Leukocyte numbers as determined by the FCM and
epigenetic
qPCR approach were highly correlated (Pearson r=0.8; p<0.0001). For the
assessment of
method comparability the inventors performed Bland-Altman analysis (17; Figure
4). The
systematic difference (bias) between the two methods was below 11% (relative)
and 18%
(absolute) for all three cell types. Moreover, non-systematic fluctuations
remained under 25%
for all three markers when comparing FACS to epigenetic qPCR, indicating low
levels of
imprecision for both approaches. According to these data, biological read-outs
of FCM and
epigenetic counting appeared to be well-correlated for all cell types. Sample
collection and
preprocessing not always warrants known volumes of blood, e.g., in dried blood
spots, barring
flow cytometric analysis. To test diagnostic accuracy of epigenetic qPCR in
these cases, the
inventors determined immune cell counts in Guthrie cards collected from 250
healthy

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
22
newborns and 30 blood cards from patients with primary immune-deficiencies
suffering from
SCID (x patients), ADA-SCID (y) and XLA (z). Upon analysis, data were
unblinded, results
were compared to data obtained with TREC and KREC analysis and the available
genetic
analysis disclosed. As shown in Figure 5, 13 out of 15 SCID cases lay out of
the 99.9%
confidence interval of the normal cohort in the CD3 to GAPDH plot, providing
for a positive
diagnosis alone. Case No 11 was out of the 99% CI in the B-cell analysis and
out of the
99.9% CI in the NK cell analysis. SCID No 10 lay inside the "normal cohort"
for the T-cell
analysis, but outside the 99.9% CI in both B-cell and NK cell to GAPDH
analyses. These
combinations in SCID cases No 10 and 11 clearly indicating a severe alteration
in the immune
cell homeostatis and would require a thorough post-screening analysis. When
analyzing
delayed onset SCID cases, No 23 was out of the 99.9% interval, No 30 out of
the 99% CI and
case No.28 appeared to be unsuspicious in the T-cell analysis. All three
cases, however, were
detected outside the 99.9% CI in the B-cell analysis and were at least outside
the 99% CI in
NK cell counts. T cell levels in XLA patients were reported outside the 99% CI
for cases No 1
and 8, but all 5 cases were outside the 99% CI in B-cells with No.1, 2, 6 and
8 outside the
99.9% CI. Also, cases 8 and 15 were outside the 99% CI for NK cells. Case No
15 was
outside the 99% CI in the B-cell analysis. Healthy controls (No 12, 14 and 29)
that were
spiked into the tests invention were within 99% CI in all assays and control
samples from
patients who had previously received stem cell transplantation (No 13, 18, 20)
were inside the
99% CI for T cells, but No 18 still identifies as "non-normal". A SCID case
with significant
maternal engraftment is not identified in this analysis, as it appears
completely unsuspicious
in all analyzed markers. Finally, the analysis of CD4 and CD8 cell fractions
support the
findings of the CD3 marker, but do not show a significant individual added
value compared to
the CD3 screening. The joint analysis of the GAPDH, CD3, B-Cell and NK cell
assay appears
to provide information for an accurate diagnosis. As the each of the three
panels is tuned to a
99% or (99.9%) CI the inventors need to correct for multiplicity and obtain
via Bonferroni
Correction a control of the family-wise-error-rate (a generalization of the
type I error) at a
level of 3% (0.3%).

CA 03051314 2019-07-23
WO 2018/146209
PCT/EP2018/053206
23
Table 1
Plasmid based controls Analyzed immune cell
populations
Cell Target
Quantifi-
type gene of Amplification cation Th cytot. B NK Mono-
Granu-
speci- qPCR system mode TpG- CpG- Cali- cells T
cells cells cells cytes locytes*
ficity assay variant variant brator CD3 CD3' CD3
CD19 CD14'
CD15'
CD4' CD8 CD56
TpG-system [#TpG] 30100 0 6443 4795 244 50 58 57 61
CpG-system [#CpG] 0 29650 8 -- 2300 7990 5100 3600 -- 5335
cu
RD!, [%] 100 0 99,8 96 06 1,1 1, 6
1,1
CD4
RDõ [%] 53,4 2,7 0,6 0,6
0,6 0,7
o EF
DD[ %] 91,4 6,1 0,6 1,1
1,4 1,3
TpG-system [#TpG] 29850 0 10457 622 5845 51 36 19 37
CpG-system [#CpG] 0 27150 6400 608 11100 7375 5720
7985
cu
CD8B RD!, [%] 100 0 8,9 90,6 0,5 0,5 0,3
0,5
ca RDt, [%] 6,9 65,1 0,6 0,4 0,2
0,4
03
EF 0,87
DD u [%] 7,3 90,6 0,4 0,4
0,3 0,5
TpG-system [#TpG] 30550 0 8723 2 2 9970 24 5 1
CpG-system [#CpG] 0 31500 4760 3205 1125 5105 3655
5790
RD!, [%] 100 0 0,0 01 89,9 0,5 01
0,0
w LRP5
ca RDt, [%] 0,0 0,0 111,0 0,3 0,1
0,0
EF 0,72
DD u [h] 0,0 0,0 91,7 0,3
0,1 0,0
TpG-system [#TpG] 27750 0 12400 150 169 170 10550 95 172
CpG-system [#CpG] 0 25750 9585 6850 16450 494
7220 11200
cu RD!, [%] 100 0 1, 5 24 1,0 95,5 1,
3 1,5
u MVD
RDt, [%] 1,7 1,9 1,9 117,5
1,1 1,9
EF 1,03
DD u [%] 1,5 2,2 1,1 101,2
1,2 1,9
TpG-system [#TpG] 33350 0 14133 12050 8320 37 59 23 28,8
CpG-system [#CpG] 0 29450 4 1 13800 9505 6810 9125,0
cu
RD!, [%] 100 0 100,0 100,0 0,3
0,6 -- 0,3 -- 0,3
.1- = CD3 D/G
RD, [%] 122,8 112,1 0,2
0,6 0,3 0,3
o FE 1,17
DD u [%] 104,4 95,2 0,2 0,5
0,3 0,2
6
CI)
5, GAPDH TpG-system [#TpG] 12050 9815 7425 15100 10110 7655 8980
u
Table 1. RD15: Relative demethylation (locus specific) in %; RD: Relative
demethylation (universal) in %; EF:
Efficiency factor; DD: Definitive demethylation (universal) in %
References as cited:
1.) Adan A, et al (2016) Flow cytometry: basic principles and applications.
Crit Rev
Biotechno1.14:1-14.
2.) Whitby L, et al. (2015) Current laboratory practices in flow cytometry for
the
enumeration of CD 4(+) T-lymphocyte subsets. Cytometry B Clin Cytom.;
88(5):305-
11
3.) Nebe CT, et al. (2013) Messunsicherheit und Qualitatssicherung im Bereich
der
Immunphanotypisierung der Lymphozytensubpopulationen im peripheren Blut. J Lab
Med 37(5):233-250.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
24
4.) Herzenberg LA, et al. (2006) Interpreting flow cytometry data. Nat.
Immunol.,
7(7):681-685.
5.) Kverneland AH, et al. (2016) Age and gender leucocytes variances and
references
values generated using the standardized ONE-Invention protocol. Cytometry A.
89(6):543-64.
6.) Maecker HT, et al. (2012) Standardizing immunophenotyping for the Human
Immunology Project. Nat Rev Immunol. 12(3):191-200.
7.) Maecker HT, McCoy JP Jr; FOCIS (2010) Human Immunophenotyping Consortium,
Amos M, et al., A model for harmonizing flow cytometry in clinical trials. Nat
Immunol. 11(11):975-8.
8.) WHO (2016). Consolidated guidelines on the use of antiretroviral drugs for
treating
and preventing HIV infection. Recommendations for a public health approach -
Second edition.
9.) European Aids Clinical Society (2015). European Guidelines for treatment
of HIV-
positive adults in Europe (Version 8.0; June 2016).
10.) Slatter MA, Cant AJ (2011) Hematopoietic stem cell transplantation for
primary immunodeficiency diseases. Ann N Y Acad Sci. 1238:122-31
11.) Thoma MD, et al. (2012) Peripheral blood lymphocyte and monocyte
recovery
and survival in acute leukemia postmyelo ablative allogeneic hematopoietic
stem cell
transplant. Biol Blood Marrow Transplant. 18(4):600-7.
12.) Nina Shah N, et al. (2015) Hematopoietic Stem Cell Transplantation for
Multiple Myeloma: Guidelines from the American Society for Blood and Marrow
Transplantation. Biol Blood Marrow Transplant 21:1155-66.
13.) Auletta JJ and Lazarus HM (2005) Immune restoration following
hematopoietic stem cell transplantation: an evolving target. Bone Marrow
Transplantation. 35:835-857.
14.) Wieczorek G, et al., (2009) Quantitative DNA methylation analysis of
FOXP3
as a new method for counting regulatory T cells in peripheral blood and solid
tissue.
Cancer Res. 69:599-608.
15.) Sehouli J, et al. (2011) Epigenetic quantification of tumor-
infiltrating T-
lymphocytes. Epigenetics 6:236-46.
16.) Baron U, et al., (2007) DNA demethylation in the human FOXP3 locus
discriminates regulatory T cells from activated FOXP3(+) conventional T cells.
Eur J
Immunol. 37:2378-2389.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
17.) Lee JW, et al., (2006) Fit-for-purpose method development and
validation for
successful biomarker measurement. Pharm Res. 23(2):312-28.15.
18.) Lewin J, et al. (2004) Quantitative DNA methylation analysis based on
four
dye trace data from direct sequencing of PCR amplificates. Bioinformatics
20:3005-
12.
19.) Mary L. McHugh (2012) Interrater reliability: the kappa statistic.
Biochem
Med (Zagreb); 22(3):276-282.
20.) Warnecke PM, Stirzaker C (1997) Detection and measurement of PCR bias
in
quantitative methylation analysis of bisulphite-treated DNA. Nucleic Acids
Res.
25(21):4422-6.
21.) de Jonge HJM, et al., (2007) Evidence based selection of housekeeping
genes.
PLoS One.2(9):e898
22.) Chen D, et al., (1999) Differential reactivity of the rat S100A4(p9Ka)
gene to
sodium bisulfite is associated with differential levels of the Si 00A4 (p9Ka)
mRNA in
rat mammary epithelial cells. J Biol Chem. 274(4):2483-91.
23.) Harrison J, Stirzaker C, Clark SJ (1998) Cytosines adjacent to
methylated CpG
sites can be partially resistant to conversion in genomic bisulfite sequencing
leading to
methylation artifacts. Anal Biochem. 264(1):129-32
24.) International Human Genome Sequencing Consortium (2001) Initial
sequencing and analysis of the human genome. Nature 409: 860-921
25.) Moore, DM, et al. (2006) CD4 percentage is an independent predictor of
survival in patients starting antiretroviral therapy with absolute CD4 cell
counts
between 200 and 350 cells/microL. HIV Med 7:383-388.
26.) Yu LM, Easterbrook PJ, Marshall T (1997) Relationship between CD4
count
and CD4% in HIV-infected people. Int J Epidemiol. 26(6):1367-72.
27.) Read SJ (2001) Recovery efficiencies of nucleic acid extraction kits
as
measured by quantitative LightCyclerTM PCR. Mol Pathol. 54(2): 86-90.
28.) Giavarina D. (2015) Understanding Bland Altman analysis. Biochemia
Medica. 25(2):141-51.
29.) Rodriguez WR, Christodoulides N, Floriano PN, Graham S, Mohanty S,
Dixon
M, Hsiang M, Peter T, Zavahir S, Thior I, Romanovicz D, Bernard B, Goodey AP,
Walker BD, McDevitt JT (2005) A microchip CD4 counting method for HIV
monitoring in resource-poor settings. PLoS Med 2(7): e182-e182.

CA 03051314 2019-07-23
WO 2018/146209 PCT/EP2018/053206
26
30.) Israeli M, Klein T, Herscovici C, Ram R, Shpilberg 0, Sredni B and
Yeshurun
M. (2013) Cellular immune function monitoring after allogeneic haematopoietic
cell
transplantation: evaluation of a new assay. Clin Exp Immunol. 172(3): 475-482.
31.) Tsikas D (2009) A proposal for comparing methods of quantitative
analysis of
endogenous compounds in biological systems by using the relative lower limit
of
quantification (rLLOQ).
J Chromatogr B Analyt Technol Biomed Life Sci. 877(23):2244-51.
32.) Boldt A, Borte S, Fricke S, Kentouche K, Emmrich F, Borte M,
Kahlenberg F,
Sack U (2014) Eight-color immunophenotyping of T-, B-, and NK-cell
subpopulations
for characterization of chronic immunodeficiencies, Cytometry B Clin Cytom.
86(3):191-206.

Representative Drawing

Sorry, the representative drawing for patent document number 3051314 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Examiner's Report 2024-05-29
Inactive: Report - No QC 2024-05-28
Amendment Received - Response to Examiner's Requisition 2023-07-07
Amendment Received - Voluntary Amendment 2023-07-07
Examiner's Report 2023-03-08
Inactive: Report - No QC 2023-03-07
Letter Sent 2022-06-23
Inactive: Single transfer 2022-05-31
Letter Sent 2022-04-06
Request for Examination Received 2022-03-01
All Requirements for Examination Determined Compliant 2022-03-01
Request for Examination Requirements Determined Compliant 2022-03-01
Common Representative Appointed 2020-11-07
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Inactive: Cover page published 2019-08-21
Inactive: Notice - National entry - No RFE 2019-08-12
Application Received - PCT 2019-08-08
Inactive: IPC assigned 2019-08-08
Inactive: IPC assigned 2019-08-08
Inactive: First IPC assigned 2019-08-08
National Entry Requirements Determined Compliant 2019-07-23
Application Published (Open to Public Inspection) 2018-08-16

Abandonment History

There is no abandonment history.

Maintenance Fee

The last payment was received on 2023-12-21

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
Basic national fee - standard 2019-07-23
MF (application, 2nd anniv.) - standard 02 2020-02-10 2020-01-20
MF (application, 3rd anniv.) - standard 03 2021-02-08 2021-02-08
MF (application, 4th anniv.) - standard 04 2022-02-08 2022-02-03
Request for examination - standard 2023-02-08 2022-03-01
Registration of a document 2022-05-31
MF (application, 5th anniv.) - standard 05 2023-02-08 2023-01-30
MF (application, 6th anniv.) - standard 06 2024-02-08 2023-12-21
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
PRECISION FOR MEDICINE GMBH
Past Owners on Record
SVEN OLEK
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2023-07-06 27 2,145
Claims 2023-07-06 3 146
Description 2019-07-22 26 1,466
Drawings 2019-07-22 9 1,109
Abstract 2019-07-22 1 43
Claims 2019-07-22 3 100
Examiner requisition 2024-05-28 3 173
Notice of National Entry 2019-08-11 1 193
Reminder of maintenance fee due 2019-10-08 1 111
Courtesy - Acknowledgement of Request for Examination 2022-04-05 1 423
Courtesy - Certificate of Recordal (Change of Name) 2022-06-22 1 387
Amendment / response to report 2023-07-06 20 928
International search report 2019-07-22 3 81
National entry request 2019-07-22 2 69
Request for examination 2022-02-28 5 119
Examiner requisition 2023-03-07 4 230